Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 58
Filter
1.
Microb Cell Fact ; 22(1): 241, 2023 Nov 27.
Article in English | MEDLINE | ID: mdl-38012629

ABSTRACT

BACKGROUND: In whole-cell bio-catalysis, the biosystems engineering paradigm shifts from the global reconfiguration of cellular metabolism as in fermentation to a more focused, and more easily modularized, optimization of comparably short cascade reactions. Human milk oligosaccharides (HMO) constitute an important field for the synthetic application of cascade bio-catalysis in resting or non-living cells. Here, we analyzed the central catalytic module for synthesis of HMO-type sialo-oligosaccharides, comprised of CMP-sialic acid synthetase (CSS) and sialyltransferase (SiaT), with the specific aim of coordinated enzyme co-expression in E. coli for reaction flux optimization in whole cell conversions producing 3'-sialyllactose (3SL). RESULTS: Difference in enzyme specific activity (CSS from Neisseria meningitidis: 36 U/mg; α2,3-SiaT from Pasteurella dagmatis: 5.7 U/mg) was compensated by differential protein co-expression from tailored plasmid constructs, giving balance between the individual activities at a high level of both (α2,3-SiaT: 9.4 × 102 U/g cell dry mass; CSS: 3.4 × 102 U/g cell dry mass). Finally, plasmid selection was guided by kinetic modeling of the coupled CSS-SiaT reactions in combination with comprehensive analytical tracking of the multistep conversion (lactose, N-acetyl neuraminic acid (Neu5Ac), cytidine 5'-triphosphate; each up to 100 mM). The half-life of SiaT in permeabilized cells (≤ 4 h) determined the efficiency of 3SL production at 37 °C. Reaction at 25 °C gave 3SL (40 ± 4 g/L) in ∼ 70% yield within 3 h, reaching a cell dry mass-specific productivity of ∼ 3 g/(g h) and avoiding intermediary CMP-Neu5Ac accumulation. CONCLUSIONS: Collectively, balanced co-expression of CSS and SiaT yields an efficient (high-flux) sialylation module to support flexible development of E. coli whole-cell catalysts for sialo-oligosaccharide production.


Subject(s)
Escherichia coli , N-Acylneuraminate Cytidylyltransferase , Humans , N-Acylneuraminate Cytidylyltransferase/genetics , N-Acylneuraminate Cytidylyltransferase/metabolism , Escherichia coli/metabolism , Oligosaccharides/metabolism , Bioengineering , Sialyltransferases/genetics , Sialyltransferases/metabolism , Catalysis
2.
Microbiol Spectr ; 11(6): e0294423, 2023 Dec 12.
Article in English | MEDLINE | ID: mdl-37850751

ABSTRACT

IMPORTANCE: The Gram-negative coccobacillus Mannheimia haemolytica is a natural inhabitant of the upper respiratory tract in ruminants and the most common bacterial agent involved in bovine respiratory disease complex development. Key virulence factors harbored by M. haemolytica are leukotoxin, lipopolysaccharide, capsule, adhesins, and neuraminidase which are involved in evading innate and adaptive immune responses. In this study, we have shown that CMP-sialic acid synthetase (neuA) is necessary for the incorporation of sialic acid onto the membrane, and inactivation of neuA results in increased phagocytosis and complement-mediated killing of M. haemolytica, thus demonstrating that sialylation contributes to the virulence of M. haemolytica.


Subject(s)
Mannheimia haemolytica , Cattle , Animals , Mannheimia haemolytica/genetics , Mannheimia haemolytica/metabolism , N-Acylneuraminate Cytidylyltransferase/genetics , N-Acylneuraminate Cytidylyltransferase/metabolism , Serogroup , Gene Deletion , Phagocytosis
3.
Proc Natl Acad Sci U S A ; 119(25): e2201129119, 2022 06 21.
Article in English | MEDLINE | ID: mdl-35696562

ABSTRACT

Sialic acids (Sias) on the B cell membrane are involved in cell migration, in the control of the complement system and, as sialic acid-binding immunoglobulin-like lectin (Siglec) ligands, in the regulation of cellular signaling. We studied the role of sialoglycans on B cells in a mouse model with B cell-specific deletion of cytidine monophosphate sialic acid synthase (CMAS), the enzyme essential for the synthesis of sialoglycans. Surprisingly, these mice showed a severe B cell deficiency in secondary lymphoid organs. Additional depletion of the complement factor C3 rescued the phenotype only marginally, demonstrating a complement-independent mechanism. The B cell survival receptor BAFF receptor was not up-regulated, and levels of activated caspase 3 and processed caspase 8 were high in B cells of Cmas-deficient mice, indicating ongoing apoptosis. Overexpressed Bcl-2 could not rescue this phenotype, pointing to extrinsic apoptosis. These results show that sialoglycans on the B cell surface are crucial for B cell survival by counteracting several death-inducing pathways.


Subject(s)
Apoptosis , B-Lymphocytes , Polysaccharides , Sialic Acids , Animals , B-Cell Activation Factor Receptor/metabolism , B-Lymphocytes/physiology , Cell Survival , Gene Deletion , Mice , N-Acylneuraminate Cytidylyltransferase/genetics , Polysaccharides/metabolism , Sialic Acid Binding Immunoglobulin-like Lectins/metabolism , Sialic Acids/metabolism
4.
Sci Rep ; 11(1): 23211, 2021 12 01.
Article in English | MEDLINE | ID: mdl-34853329

ABSTRACT

Vertebrate CMP-sialic acid synthetase (CSS), which catalyzes the synthesis of CMP-sialic acid (CMP-Sia), consists of a 28 kDa-N-domain and a 20 kDa-C-domain. The N-domain is known to be a catalytic domain; however, the significance of the C-domain still remains unknown. To elucidate the function of the C-domain at the organism level, we screened the medaka TILLING library and obtained medaka with non-synonymous mutations (t911a), or single amino acid substitutions of CSS, L304Q, in the C-domain. Prominently, most L304Q medaka was lethal within 19 days post-fertilization (dpf). L304Q young fry displayed free Sia accumulation, and impairment of sialylation, up to 8 dpf. At 8 dpf, a marked abnormality in ventricular contraction and skeletal myogenesis was observed. To gain insight into the mechanism of L304Q-induced abnormalities, L304Q was biochemically characterized. Although bacterially expressed soluble L304Q and WT showed the similar Vmax/Km values, very few soluble L304Q was detected when expressed in CHO cells in sharp contrast to the WT. Additionally, the thermostability of various mutations of L304 greatly decreased, except for WT and L304I. These results suggest that L304 is important for the stability of CSS, and that an appropriate level of expression of soluble CSS is significant for animal survival.


Subject(s)
Fish Diseases/genetics , Fish Proteins/genetics , N-Acylneuraminate Cytidylyltransferase/genetics , Oryzias/genetics , Point Mutation , Animals , CHO Cells , Cardiomyopathies/genetics , Cardiomyopathies/veterinary , Cricetulus , Enzyme Stability , Fish Proteins/chemistry , Models, Molecular , N-Acylneuraminate Cytidylyltransferase/chemistry , Oryzias/physiology , Protein Domains , Solubility
5.
Int J Mol Sci ; 22(11)2021 Jun 04.
Article in English | MEDLINE | ID: mdl-34200006

ABSTRACT

Influenza A viruses (IAVs) initiate infection by attaching Hemagglutinin (HA) on the viral envelope to sialic acid (SA) receptors on the cell surface. Importantly, HA of human IAVs has a higher affinity for α-2,6-linked SA receptors, and avian strains prefer α-2,3-linked SA receptors, whereas swine strains have a strong affinity for both SA receptors. Host gene CMAS and ST3GAL4 were found to be essential for IAV attachment and entry. Loss of CMAS and ST3GAL4 hindered the synthesis of sialic acid receptors, which in turn prevented the adsorption of IAV. Further, the knockout of CMAS had an effect on the adsorption of swine, avian and human IAVs. However, ST3GAL4 knockout prevented the adsorption of swine and avian IAV and the impact on avian IAV was more distinct, whereas it had no effect on the adsorption of human IAV. Collectively, our findings demonstrate that knocking out CMAS and ST3GAL4 negatively regulated IAV replication by inhibiting the synthesis of SA receptors, which also provides new insights into the production of gene-edited animals in the future.


Subject(s)
Influenza A virus/physiology , N-Acylneuraminate Cytidylyltransferase/antagonists & inhibitors , Orthomyxoviridae Infections/virology , Receptors, Cell Surface/metabolism , Sialyltransferases/antagonists & inhibitors , Virus Replication , Animals , CRISPR-Cas Systems , N-Acetylneuraminic Acid/metabolism , N-Acylneuraminate Cytidylyltransferase/genetics , N-Acylneuraminate Cytidylyltransferase/metabolism , Orthomyxoviridae Infections/metabolism , Orthomyxoviridae Infections/pathology , Swine
6.
J Virol ; 95(2)2020 12 22.
Article in English | MEDLINE | ID: mdl-33087464

ABSTRACT

Engagement of cell surface receptors by viruses is a critical determinant of viral tropism and disease. The reovirus attachment protein σ1 binds sialylated glycans and proteinaceous receptors to mediate infection, but the specific requirements for different cell types are not entirely known. To identify host factors required for reovirus-induced cell death, we conducted a CRISPR-knockout screen targeting over 20,000 genes in murine microglial BV2 cells. Candidate genes required for reovirus to cause cell death were highly enriched for sialic acid synthesis and transport. Two of the top candidates identified, CMP N-acetylneuraminic acid synthetase (Cmas) and solute carrier family 35 member A1 (Slc35a1), promote sialic acid expression on the cell surface. Two reovirus strains that differ in the capacity to bind sialic acid, T3SA+ and T3SA-, were used to evaluate Cmas and Slc35a1 as potential host genes required for reovirus infection. Following CRISPR-Cas9 disruption of either gene, cell surface expression of sialic acid was diminished. These results correlated with decreased binding of strain T3SA+, which is capable of engaging sialic acid. Disruption of either gene did not alter the low-level binding of T3SA-, which does not engage sialic acid. Furthermore, infectivity of T3SA+ was diminished to levels similar to those of T3SA- in cells lacking Cmas and Slc35a1 by CRISPR ablation. However, exogenous expression of Cmas and Slc35a1 into the respective null cells restored sialic acid expression and T3SA+ binding and infectivity. These results demonstrate that Cmas and Slc35a1, which mediate cell surface expression of sialic acid, are required in murine microglial cells for efficient reovirus binding and infection.IMPORTANCE Attachment factors and receptors are important determinants of dissemination and tropism during reovirus-induced disease. In a CRISPR cell survival screen, we discovered two genes, Cmas and Slc35a1, which encode proteins required for sialic acid expression on the cell surface and mediate reovirus infection of microglial cells. This work elucidates host genes that render microglial cells susceptible to reovirus infection and expands current understanding of the receptors on microglial cells that are engaged by reovirus. Such knowledge may lead to new strategies to selectively target microglial cells for oncolytic applications.


Subject(s)
N-Acylneuraminate Cytidylyltransferase/metabolism , Nucleotide Transport Proteins/metabolism , Reoviridae Infections/virology , Reoviridae/physiology , Animals , Capsid Proteins/genetics , Capsid Proteins/metabolism , Cell Line , Cell Membrane/metabolism , Cell Survival , Mice , N-Acetylneuraminic Acid/metabolism , N-Acylneuraminate Cytidylyltransferase/genetics , Nucleotide Transport Proteins/genetics , Receptors, Virus/metabolism , Reoviridae/genetics , Reoviridae/metabolism , Reoviridae Infections/metabolism , Virus Attachment , Virus Replication
7.
Biochemistry ; 59(34): 3157-3168, 2020 09 01.
Article in English | MEDLINE | ID: mdl-31583886

ABSTRACT

Cytidine 5'-monophosphate (CMP)-sialic acid synthetase (CSS) is an essential enzyme involved in the biosynthesis of carbohydrates and glycoconjugates containing sialic acids, a class of α-keto acids that are generally terminal key recognition residues by many proteins that play important biological and pathological roles. The CSS from Neisseria meningitidis (NmCSS) has been commonly used with other enzymes such as sialic acid aldolase and/or sialyltransferase in synthesizing a diverse array of compounds containing sialic acid or its naturally occurring and non-natural derivatives. To better understand its catalytic mechanism and substrate promiscuity, four NmCSS crystal structures trapped at various stages of the catalytic cycle with bound substrates, substrate analogues, and products have been obtained and are presented here. These structures suggest a mechanism for an "open" and "closed" conformational transition that occurs as sialic acid binds to the NmCSS/cytidine-5'-triphosphate (CTP) complex. The closed conformation positions critical residues to help facilitate the nucleophilic attack of sialic acid C2-OH to the α-phosphate of CTP, which is also aided by two observed divalent cations. Product formation drives the active site opening, promoting the release of products.


Subject(s)
Biocatalysis , N-Acylneuraminate Cytidylyltransferase/chemistry , N-Acylneuraminate Cytidylyltransferase/metabolism , Neisseria meningitidis/enzymology , Catalytic Domain , Crystallography, X-Ray , Models, Molecular , Mutation , N-Acylneuraminate Cytidylyltransferase/genetics
8.
Ann Hum Genet ; 84(1): 46-53, 2020 01.
Article in English | MEDLINE | ID: mdl-31495922

ABSTRACT

Intellectual disability (ID) describes a wide range of serious human diseases caused by defects in central nervous system development and function. Some mutant genes have been found to be associated with these diseases, but not all cases can be explained, thus suggesting that other disease-causing genes have not yet been discovered. Sialic acid is involved in a number of key biological processes, including embryo formation, nerve cell growth, and cancer cell metastasis, and very recently it has been suggested that N-acetylneuraminic acid synthase-mediated synthesis of sialic acid is required for brain and skeletal development. CMP-sialic acid synthetase (CMAS) is one of four enzymes involved in NeuNAc metabolism, as it catalyzes the formation of CMP-NeuNAc. Before the present study, no links between mutations in CMAS and incidences of human ID had been reported. In the current study, we recruited a recessive nonsyndromic ID pedigree with consanguineous marriage in which all patients have typical clinical manifestations of ID. We identified the NM_018686.3:c.563G > A (p.Arg188His) substitution in CMAS as being responsible for the disease in this family. Conservation analysis, structural prediction, and enzyme activity experiments demonstrated that (p.Arg188His) influences protein dimerization and alters CMAS enzyme activity. Our results offer a new orientation for future research and clinical diagnosis.


Subject(s)
Genes, Recessive , Homozygote , Intellectual Disability/etiology , Mutation , N-Acylneuraminate Cytidylyltransferase/genetics , Adult , Amino Acid Sequence , Consanguinity , Female , Follow-Up Studies , Humans , Intellectual Disability/pathology , Male , Middle Aged , Pedigree , Prognosis , Sequence Homology , Young Adult
9.
Nat Commun ; 10(1): 3698, 2019 08 16.
Article in English | MEDLINE | ID: mdl-31420548

ABSTRACT

Phosphonates are rare and unusually bioactive natural products. However, most bacterial phosphonate biosynthetic capacity is dedicated to tailoring cell surfaces with molecules like 2-aminoethylphosphonate (AEP). Although phosphoenolpyruvate mutase (Ppm)-catalyzed installation of C-P bonds is known, subsequent phosphonyl tailoring (Pnt) pathway steps remain enigmatic. Here we identify nucleotidyltransferases in over two-thirds of phosphonate biosynthetic gene clusters, including direct fusions to ~60% of Ppm enzymes. We characterize two putative phosphonyl tailoring cytidylyltransferases (PntCs) that prefer AEP over phosphocholine (P-Cho) - a similar substrate used by the related enzyme LicC, which is a virulence factor in Streptococcus pneumoniae. PntC structural analyses reveal steric discrimination against phosphocholine. These findings highlight nucleotidyl activation as a predominant chemical logic in phosphonate biosynthesis and set the stage for probing diverse phosphonyl tailoring pathways.


Subject(s)
Aminoethylphosphonic Acid/metabolism , Bacteria/metabolism , Bacterial Proteins/metabolism , Biosynthetic Pathways/physiology , N-Acylneuraminate Cytidylyltransferase/metabolism , Organophosphonates/metabolism , Actinobacteria , Bacteria/genetics , Bacterial Proteins/genetics , Cell Wall/metabolism , Crystallization , Crystallography, X-Ray , Escherichia coli , N-Acylneuraminate Cytidylyltransferase/genetics , Nucleotidyltransferases/genetics , Nucleotidyltransferases/metabolism , Phospholipids/metabolism , Phosphorylcholine/metabolism , Phosphotransferases (Phosphomutases) , Polysaccharides/metabolism , Substrate Specificity
10.
Carbohydr Res ; 472: 115-121, 2019 Jan 15.
Article in English | MEDLINE | ID: mdl-30562693

ABSTRACT

An efficient streamlined chemoenzymatic approach has been developed for gram-scale synthesis of Lewis a angtigen (LeaßProN3) and a library of sialyl Lewis a antigens (sLeaßProN3) containing different sialic acid forms. Intially, commercially available inexpensive N-acetylglucosamine (GlcNAc) was converted to its N'-glycosyl p-toluenesulfonohydrazide in one step. Followed by chemical glycosylation, GlcNAcßProN3 was synthesized using this protecting group-free method in high yield (82%). Sequential one-pot multienzyme (OPME) ß1-3-galactosylation of GlcNAcßProN3 followed by OPME α1-4-fucosylation reactions produced target LeaßProN3 in gram-scale. Structurally diverse sialic acid forms was successfully introduced using a OPME sialylation reation containing a CMP-sialic acid synthetase and Pasteurella multocida α2-3-sialyltransferase 1 (PmST1) mutant PmST1 M144D with or without a sialic acid aldolase to form sLeaßProN3 containing naturally occurring or non-natural sialic acid forms in preparative scales.


Subject(s)
Lewis Blood Group Antigens/chemistry , N-Acylneuraminate Cytidylyltransferase/metabolism , Sialic Acids/chemistry , Sialyltransferases/metabolism , Acetylglucosamine/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Mutation , N-Acylneuraminate Cytidylyltransferase/genetics , Pasteurella multocida/enzymology , Sialyltransferases/genetics , Tosyl Compounds/chemistry
11.
Int J Cancer ; 144(9): 2290-2302, 2019 05 01.
Article in English | MEDLINE | ID: mdl-30578646

ABSTRACT

Sialylated glycan structures are known for their immunomodulatory capacities and their contribution to tumor immune evasion. However, the role of aberrant sialylation in colorectal cancer and the consequences of complete tumor desialylation on anti-tumor immunity remain unstudied. Here, we report that CRISPR/Cas9-mediated knock out of the CMAS gene, encoding a key enzyme in the sialylation pathway, in the mouse colorectal cancer MC38 cell line completely abrogated cell surface expression of sialic acids (MC38-Sianull ) and, unexpectedly, significantly increased in vivo tumor growth compared to the control MC38-MOCK cells. This enhanced tumor growth of MC38-Sianull cells could be attributed to decreased CD8+ T cell frequencies in the tumor microenvironment only, as immune cell frequencies in tumor-draining lymph nodes remained unaffected. In addition, MC38-Sianull cells were able to induce CD8+ T cell apoptosis in an antigen-independent manner. Moreover, low CMAS gene expression correlated with reduced recurrence-free survival in a human colorectal cancer cohort, supporting the clinical relevance of our work. Together, these results demonstrate for the first time a detrimental effect of complete tumor desialylation on colorectal cancer tumor growth, which greatly impacts the design of novel cancer therapeutics aimed at altering the tumor glycosylation profile.


Subject(s)
Apoptosis/immunology , CD8-Positive T-Lymphocytes/immunology , Colorectal Neoplasms/pathology , N-Acylneuraminate Cytidylyltransferase/genetics , Sialic Acids/metabolism , Tumor Escape/immunology , Animals , CRISPR-Cas Systems/genetics , Cell Line, Tumor , Colorectal Neoplasms/genetics , Disease-Free Survival , Glycosylation , Humans , Lymphocyte Count , Mice , Mice, Inbred C57BL , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology
12.
Chembiochem ; 18(13): 1251-1259, 2017 07 04.
Article in English | MEDLINE | ID: mdl-28395125

ABSTRACT

Sialylation of glycoproteins and glycolipids is catalyzed by sialyltransferases in the Golgi of mammalian cells, whereby sialic acid residues are added at the nonreducing ends of oligosaccharides. Because sialylated glycans play critical roles in a number of human physio-pathological processes, the past two decades have witnessed the development of modified sialic acid derivatives for a better understanding of sialic acid biology and for the development of new therapeutic targets. However, nothing is known about how individual mammalian sialyltransferases tolerate and behave towards these unnatural CMP-sialic acid donors. In this study, we devised several approaches to investigate the donor specificity of the human ß-d-galactoside sialyltransferases ST6Gal I and ST3Gal I by using two CMP-sialic acids: CMP-Neu5Ac, and CMP-Neu5N-(4pentynoyl)neuraminic acid (CMP-SiaNAl), an unnatural CMP-sialic acid donor with an extended and functionalized N-acyl moiety.


Subject(s)
Antigens, CD/metabolism , Cytidine Monophosphate N-Acetylneuraminic Acid/metabolism , Cytidine Monophosphate/analogs & derivatives , Glycolipids/metabolism , Glycoproteins/metabolism , Polysaccharides/metabolism , Sialic Acids/metabolism , Sialyltransferases/metabolism , Antigens, CD/chemistry , Antigens, CD/genetics , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Cloning, Molecular , Cytidine Monophosphate/chemistry , Cytidine Monophosphate/metabolism , Cytidine Monophosphate N-Acetylneuraminic Acid/chemistry , Gene Expression , Glycolipids/chemistry , Glycoproteins/chemistry , Glycoproteins/genetics , Glycosylation , HEK293 Cells , Humans , Kinetics , N-Acylneuraminate Cytidylyltransferase/genetics , N-Acylneuraminate Cytidylyltransferase/metabolism , Neisseria meningitidis/chemistry , Neisseria meningitidis/enzymology , Polysaccharides/chemistry , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sialic Acids/chemistry , Sialyltransferases/chemistry , Sialyltransferases/genetics , Substrate Specificity , beta-Galactoside alpha-2,3-Sialyltransferase
13.
Chembiochem ; 18(13): 1305-1316, 2017 07 04.
Article in English | MEDLINE | ID: mdl-28374933

ABSTRACT

The negatively charged nonulose sialic acid (Sia) is essential for murine development in vivo. In order to elucidate the impact of sialylation on differentiation processes in the absence of maternal influences, we generated mouse embryonic stem cell (mESC) lines that lack CMP-Sia synthetase (CMAS) and thereby the ability to activate Sia to CMP-Sia. Loss of CMAS activity resulted in an asialo cell surface accompanied by an increase in glycoconjugates with terminal galactosyl and oligo-LacNAc residues, as well as intracellular accumulation of free Sia. Remarkably, these changes did not impact intracellular metabolites or the morphology and transcriptome of pluripotent mESC lines. Moreover, the capacity of Cmas-/- mESCs for undirected differentiation into embryoid bodies, germ layer formation and even the generation of beating cardiomyocytes provides first and conclusive evidence that pluripotency and differentiation of mESC in vitro can proceed in the absence of (poly)sialoglycans.


Subject(s)
Germ Layers/metabolism , Mouse Embryonic Stem Cells/metabolism , Myocytes, Cardiac/metabolism , N-Acylneuraminate Cytidylyltransferase/deficiency , Pluripotent Stem Cells/metabolism , Sialic Acids/metabolism , Amino Sugars/metabolism , Animals , Cell Differentiation , Cell Line , Embryo, Mammalian , Embryoid Bodies/cytology , Embryoid Bodies/metabolism , Founder Effect , Galactose/metabolism , Gene Expression , Germ Layers/cytology , Glycoconjugates/metabolism , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mouse Embryonic Stem Cells/cytology , Myocytes, Cardiac/cytology , N-Acylneuraminate Cytidylyltransferase/genetics , Pluripotent Stem Cells/cytology , Transcriptome
14.
Glycobiology ; 27(4): 329-341, 2017 04 01.
Article in English | MEDLINE | ID: mdl-27986833

ABSTRACT

The occurrence and biological importance of sialic acid (Sia) and its metabolic enzymes in insects have been studied using Drosophila melanogaster. The most prominent feature of D. melanogaster CMP-Sia synthetase (DmCSS) is its Golgi-localization, contrasted with nuclear localization of vertebrate CSSs. However, it remains unclear if the Golgi-localization is common to other insect CSSs and why it happens. To answer these questions, Aedes aegypti (mosquito) CSS (AaCSS) and Tribolium castaneum (beetle) CSS (TcCSS) were cloned and characterized for their activity and subcellular localization. Our new findings show: (1) AaCSS and TcCSS share a common overall structure with DmCSS in terms of evolutionarily conserved motifs and the absence of the C-terminal domain typical to vertebrate CSSs; (2) when expressed in mammalian and insect cells, AaCSS and TcCSS showed in vivo and in vitro CSS activities, similar to DmCSS. In contrast, when expressed in bacteria, they lacked CSS activity because the N-terminal hydrophobic region appeared to induce protein aggregation; (3) when expressed in Drosophila S2 cells, AaCSS and TcCSS were predominantly localized in the ER, but not in the Golgi. Surprisingly, DmCSS was mainly secreted into the culture medium, although partially detected in Golgi. Consistent with these results, the N-terminal hydrophobic regions of AaCSS and TcCSS functioned as a signal peptide to render them soluble in the ER, while the N-terminus of DmCSS functioned as a membrane-spanning region of type II transmembrane proteins whose cytosolic KLK sequence functioned as an ER export signal. Accordingly, the differential subcellular localization of insect CSSs are distinctively more diverse than previously recognized.


Subject(s)
N-Acetylneuraminic Acid/genetics , N-Acylneuraminate Cytidylyltransferase/chemistry , N-Acylneuraminate Cytidylyltransferase/genetics , Aedes/enzymology , Amino Acid Motifs/genetics , Animals , Drosophila melanogaster/enzymology , Drosophila melanogaster/genetics , Golgi Apparatus/enzymology , Golgi Apparatus/genetics , Mutation , N-Acetylneuraminic Acid/chemistry , N-Acetylneuraminic Acid/metabolism , N-Acylneuraminate Cytidylyltransferase/metabolism , Protein Conformation , Tribolium/enzymology
15.
ACS Chem Biol ; 11(8): 2131-9, 2016 08 19.
Article in English | MEDLINE | ID: mdl-27380425

ABSTRACT

Many mechanisms have been proposed for how heightened aerobic glycolytic metabolism fuels cancer pathogenicity, but there are still many unexplored pathways. Here, we have performed metabolomic profiling to map glucose incorporation into metabolic pathways upon transformation of mammary epithelial cells by 11 commonly mutated human oncogenes. We show that transformation of mammary epithelial cells by oncogenic stimuli commonly shunts glucose-derived carbons into synthesis of sialic acid, a hexosamine pathway metabolite that is converted to CMP-sialic acid by cytidine monophosphate N-acetylneuraminic acid synthase (CMAS) as a precursor to glycoprotein and glycolipid sialylation. We show that CMAS knockdown leads to elevations in intracellular sialic acid levels, a depletion of cellular sialylation, and alterations in the expression of many cancer-relevant genes to impair breast cancer pathogenicity. Our study reveals the heretofore unrecognized role of sialic acid metabolism and protein sialylation in regulating the expression of genes that maintain breast cancer pathogenicity.


Subject(s)
Breast Neoplasms/metabolism , Gene Expression Regulation, Neoplastic , N-Acetylneuraminic Acid/metabolism , Neoplasm Proteins/metabolism , Animals , Breast Neoplasms/enzymology , Breast Neoplasms/pathology , Cell Line, Tumor , Female , Gene Knockdown Techniques , Heterografts , Humans , Metabolomics , Mice, SCID , N-Acylneuraminate Cytidylyltransferase/genetics , N-Acylneuraminate Cytidylyltransferase/metabolism , Transcriptome
16.
Biochem J ; 473(13): 1905-16, 2016 07 01.
Article in English | MEDLINE | ID: mdl-27114558

ABSTRACT

CMP-sialic acid synthetase (CSAS) is a key enzyme of the sialylation pathway. CSAS produces the activated sugar donor, CMP-sialic acid, which serves as a substrate for sialyltransferases to modify glycan termini with sialic acid. Unlike other animal CSASs that normally localize in the nucleus, Drosophila melanogaster CSAS (DmCSAS) localizes in the cell secretory compartment, predominantly in the Golgi, which suggests that this enzyme has properties distinct from those of its vertebrate counterparts. To test this hypothesis, we purified recombinant DmCSAS and characterized its activity in vitro Our experiments revealed several unique features of this enzyme. DmCSAS displays specificity for N-acetylneuraminic acid as a substrate, shows preference for lower pH and can function with a broad range of metal cofactors. When tested at a pH corresponding to the Golgi compartment, the enzyme showed significant activity with several metal cations, including Zn(2+), Fe(2+), Co(2+) and Mn(2+), whereas the activity with Mg(2+) was found to be low. Protein sequence analysis and site-specific mutagenesis identified an aspartic acid residue that is necessary for enzymatic activity and predicted to be involved in co-ordinating a metal cofactor. DmCSAS enzymatic activity was found to be essential in vivo for rescuing the phenotype of DmCSAS mutants. Finally, our experiments revealed a steep dependence of the enzymatic activity on temperature. Taken together, our results indicate that DmCSAS underwent evolutionary adaptation to pH and ionic environment different from that of counterpart synthetases in vertebrates. Our data also suggest that environmental temperatures can regulate Drosophila sialylation, thus modulating neural transmission.


Subject(s)
Drosophila Proteins/chemistry , Drosophila Proteins/metabolism , N-Acylneuraminate Cytidylyltransferase/chemistry , N-Acylneuraminate Cytidylyltransferase/metabolism , Amino Acid Sequence , Animals , Aspartic Acid/chemistry , Aspartic Acid/metabolism , Cells, Cultured , Drosophila , Drosophila Proteins/classification , Drosophila Proteins/genetics , Humans , Hydrogen-Ion Concentration , Kinetics , Magnesium/metabolism , Manganese/metabolism , Mutation , N-Acylneuraminate Cytidylyltransferase/classification , N-Acylneuraminate Cytidylyltransferase/genetics , Phylogeny , Structure-Activity Relationship , Substrate Specificity , Temperature
17.
Clin Microbiol Infect ; 20(7): O435-41, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24245827

ABSTRACT

Sequence-based typing (SBT) combined with monoclonal antibody subgrouping of Legionella pneumophila isolates is at present considered to be the reference standard during epidemiological investigation of Legionnaires' disease outbreaks. In some isolates of L. pneumophila, the seventh allele of the standard SBT scheme, neuA, is not amplified, because a homologue that is refractory to amplification with the standard neuA primers is present. Consequently, a complete seven-allele profile, and hence a sequence type, cannot be obtained. Subsequently, primers were designed to amplify both neuA and the homologue, but these yielded suboptimal sequencing results. In this study, novel primers specific for the neuA homologue were designed and internationally validated by members of the ESCMID Study Group for Legionella Infections at national and regional Legionella reference laboratories with a modified version of the online L. pneumophila sequence quality tool. To date, the addition of the neuAh target to the SBT protocol has allowed full typing data to be obtained for 108 isolates of 11 different serogroups, namely 1, 2, 3, 4, 5, 6, 7, 8, 10, 13, and 14, which could not previously be typed with the standard SBT neuA primers. Further studies are necessary to determine why it is still not possible to obtain either a neuA or a neuAh allele from three serogroup 11 isolates.


Subject(s)
Legionella pneumophila/classification , Legionella pneumophila/genetics , Molecular Typing/methods , N-Acylneuraminate Cytidylyltransferase/genetics , DNA, Bacterial/chemistry , DNA, Bacterial/genetics , Disease Outbreaks , Humans , Legionella pneumophila/enzymology , Legionnaires' Disease/epidemiology , Legionnaires' Disease/microbiology , Molecular Epidemiology/methods , Molecular Sequence Data , Sequence Analysis, DNA
18.
J Neurosci ; 33(30): 12306-15, 2013 Jul 24.
Article in English | MEDLINE | ID: mdl-23884937

ABSTRACT

While sialylation plays important functions in the nervous system, the complexity of glycosylation pathways and limitations of genetic approaches preclude the efficient analysis of these functions in mammalian organisms. Drosophila has recently emerged as a promising model for studying neural sialylation. Drosophila sialyltransferase, DSiaT, was shown to be involved in the regulation of neural transmission. However, the sialylation pathway was not investigated in Drosophila beyond the DSiaT-mediated step. Here we focused on the function of Drosophila cytidine monophosphate-sialic acid synthetase (CSAS), the enzyme providing a sugar donor for DSiaT. Our results revealed that the expression of CSAS is tightly regulated and restricted to the CNS throughout development and in adult flies. We generated CSAS mutants and analyzed their phenotypes using behavioral and physiological approaches. Our experiments demonstrated that mutant phenotypes of CSAS are similar to those of DSiaT, including decreased longevity, temperature-induced paralysis, locomotor abnormalities, and defects of neural transmission at neuromuscular junctions. Genetic interactions between CSAS, DSiaT, and voltage-gated channel genes paralytic and seizure were consistent with the hypothesis that CSAS and DSiaT function within the same pathway regulating neural excitability. Intriguingly, these interactions also suggested that CSAS and DSiaT have some additional, independent functions. Moreover, unlike its mammalian counterparts that work in the nucleus, Drosophila CSAS was found to be a glycoprotein-bearing N-glycans and predominantly localized in vivo to the Golgi compartment. Our work provides the first systematic analysis of in vivo functions of a eukaryotic CSAS gene and sheds light on evolutionary relationships among metazoan CSAS proteins.


Subject(s)
Cytidine Monophosphate/metabolism , Drosophila Proteins/genetics , Drosophila/enzymology , Ligases/genetics , N-Acetylneuraminic Acid/metabolism , N-Acylneuraminate Cytidylyltransferase/genetics , Nervous System Physiological Phenomena/genetics , Animals , Animals, Genetically Modified , Drosophila/genetics , Drosophila Proteins/metabolism , Evolution, Molecular , Gene Expression Regulation, Developmental/physiology , Ligases/metabolism , Longevity/genetics , N-Acylneuraminate Cytidylyltransferase/metabolism , Neuromuscular Junction/genetics , Neuromuscular Junction/metabolism , Paralysis/genetics , Paralysis/metabolism , Secretory Vesicles/physiology , Sialyltransferases/genetics , Sialyltransferases/metabolism , Synaptic Transmission/genetics , Synaptic Transmission/physiology , Temperature
19.
J Am Soc Nephrol ; 23(8): 1319-28, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22745475

ABSTRACT

The role of sialylation in kidney biology is not fully understood. The synthesis of sialoglycoconjugates, which form the outermost structures of animal cells, requires CMP-sialic acid, which is a product of the nuclear enzyme CMAS. We used a knock-in strategy to create a mouse with point mutations in the canonical nuclear localization signal of CMAS, which relocated the enzyme to the cytoplasm of transfected cells without affecting its activity. Although insufficient to prevent nuclear entry in mice, the mutation led to a drastically reduced concentration of nuclear-expressed enzyme. Mice homozygous for the mutation died from kidney failure within 72 hours after birth. The Cmas(nls) mouse exhibited podocyte foot process effacement, absence of slit diaphragms, and massive proteinuria, recapitulating features of nephrin-knockout mice and of patients with Finnish-type congenital nephrotic syndrome. Although the Cmas(nls) mouse displayed normal sialylation in all organs including kidney, a critical shortage of CMP-sialic acid prevented sialylation of nephrin and podocalyxin in the maturing podocyte where it is required during the formation of foot processes. Accordingly, the sialylation defects progressed with time and paralleled the morphologic changes. In summary, sialylation is critical during the development of the glomerular filtration barrier and required for the proper function of nephrin. Whether altered sialylation impairs nephrin function in human disease requires further study.


Subject(s)
Glomerular Filtration Barrier/embryology , Membrane Proteins/metabolism , N-Acetylneuraminic Acid/metabolism , N-Acylneuraminate Cytidylyltransferase/metabolism , Podocytes/physiology , Animals , Cell Nucleus/metabolism , Gene Knock-In Techniques , Mice , Mice, Inbred C57BL , N-Acylneuraminate Cytidylyltransferase/genetics , Phenotype , Podocytes/ultrastructure , Sialoglycoproteins/metabolism
20.
J Biol Chem ; 287(16): 13239-48, 2012 Apr 13.
Article in English | MEDLINE | ID: mdl-22351762

ABSTRACT

Sialic acids (Sia) form the nonreducing end of the bulk of cell surface-expressed glycoconjugates. They are, therefore, major elements in intercellular communication processes. The addition of Sia to glycoconjugates requires metabolic activation to CMP-Sia, catalyzed by CMP-Sia synthetase (CMAS). This highly conserved enzyme is located in the cell nucleus in all vertebrates investigated to date, but its nuclear function remains elusive. Here, we describe the identification and characterization of two Cmas enzymes in Danio rerio (dreCmas), one of which is exclusively localized in the cytosol. We show that the two cmas genes most likely originated from the third whole genome duplication, which occurred at the base of teleost radiation. cmas paralogues were maintained in fishes of the Otocephala clade, whereas one copy got subsequently lost in Euteleostei (e.g. rainbow trout). In zebrafish, the two genes exhibited a distinct spatial expression pattern. The products of these genes (dreCmas1 and dreCmas2) diverged not only with respect to subcellular localization but also in substrate specificity. Nuclear dreCmas1 favored N-acetylneuraminic acid, whereas the cytosolic dreCmas2 showed highest affinity for 5-deamino-neuraminic acid. The subcellular localization was confirmed for the endogenous enzymes in fractionated zebrafish lysates. Nuclear entry of dreCmas1 was mediated by a bipartite nuclear localization signal, which seemed irrelevant for other enzymatic functions. With the current demonstration that in zebrafish two subfunctionalized cmas paralogues co-exist, we introduce a novel and unique model to detail the roles that CMAS has in the nucleus and in the sialylation pathways of animal cells.


Subject(s)
Evolution, Molecular , N-Acylneuraminate Cytidylyltransferase/genetics , Zebrafish/genetics , Amino Acid Sequence , Animals , Cell Line, Tumor , Cell Nucleus/enzymology , Gene Expression Regulation, Developmental/physiology , Gene Expression Regulation, Enzymologic/physiology , Glycosylation , Mice , Molecular Sequence Data , N-Acetylneuraminic Acid/metabolism , N-Acylneuraminate Cytidylyltransferase/chemistry , N-Acylneuraminate Cytidylyltransferase/metabolism , NIH 3T3 Cells , RNA, Messenger/genetics , Substrate Specificity/physiology , Zebrafish/embryology
SELECTION OF CITATIONS
SEARCH DETAIL
...